1
|
Shin YS, Yun JK, Jung J, Park JH, Song SY, Choi EK, Choi S, Kim HR, Kim YH, Kim DK, Lee GD, Yoon SM. Comparison of Metastasectomy and Stereotactic Body Radiation Therapy for Pulmonary Oligometastasis From Hepatocellular Carcinoma: A Propensity Score-Weighted Analysis. Int J Radiat Oncol Biol Phys 2025; 121:432-441. [PMID: 39299553 DOI: 10.1016/j.ijrobp.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE Although there is a growing role for local therapy in patients with hepatocellular carcinoma (HCC) and pulmonary oligometastasis, it remains unclear whether metastatectomy or stereotactic body radiation therapy (SBRT) is the more effective treatment for these patients. We aimed to compare the oncologic outcomes of metastasectomy and SBRT for HCC with pulmonary oligometastasis. METHODS AND MATERIALS We retrospectively analyzed 209 patients with HCC with 322 metastatic lung lesions who underwent either metastasectomy (150 patients with 241 lesions) or SBRT (59 patients with 81 lesions) between January 2008 and December 2018. Propensity score-based inverse probability of treatment weighting was used to minimize potential bias between the 2 groups. RESULTS The median follow-up duration was 39.8 months (range, 2.3-166.9 months). The 2-year rate of freedom from local progression was 98.2% in the metastasectomy group and 97.0% in the SBRT group (P = .197). The 2-year rates of overt systemic progression-free survival (PFS) (51.0% vs 46.1%; P = .274), PFS (26.3% vs 9.1%; P = .074), and overall survival (OS, 74.0% vs 57.6%; P = .006) were higher in the metastasectomy group. After the probability of treatment weighting adjustment, the 2-year rates of overt systemic PFS (50.8% vs 52.7%; P = .396), PFS (23.0% vs 24.7%; P = .478), and OS (72.6% vs 83.0%, P = .428) were not significantly different between the 2 groups. In multivariate analysis, viable intrahepatic lesions and the number of prior liver-directed therapies were found to be significant prognostic factors for OS and PFS. The time interval between HCC diagnosis and the development of pulmonary metastases was also significantly associated with OS. CONCLUSIONS Both metastasectomy and SBRT demonstrated excellent local control and comparable oncologic outcomes in patients with pulmonary oligometastasis from HCC. The treatment modality for these patients could be determined based on the individual patient's condition and intrahepatic disease status.
Collapse
Affiliation(s)
| | - Jae Kwang Yun
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | - Sehoon Choi
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyeong Ryul Kim
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong-Hee Kim
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong Kwan Kim
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Geun Dong Lee
- Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | | |
Collapse
|
2
|
He SQ, Liu GY, Yu YH, Wang L, Zhang GY, Peng DS, Bei WX, Chen CL, Lv SH, Zhao ZY, Huang Y, Xiang YQ. Efficacy of local-regional radiotherapy in de novo metastatic nasopharyngeal carcinoma patients receiving chemo-immunotherapy: A multicenter, propensity score matching study. Radiother Oncol 2025; 203:110687. [PMID: 39709030 DOI: 10.1016/j.radonc.2024.110687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND To evaluate the efficacy of local-regional radiotherapy (LRRT) in de novo metastatic nasopharyngeal carcinoma (dm NPC) patients receiving chemo-immunotherapy as first-line treatment and select the beneficiaries from LRRT. METHODS AND MATERIALS M1-NPC patients receiving platinum-based chemo-immunotherapy with or without LRRT from four centers were included in this study. The propensity score matching (PSM) analysis was employed to balance the baseline characteristics between the LRRT and non-LRRT groups. RESULTS 546 dm NPC patients (140 patients in the non-LRRT group and 406 patients in the LRRT group) were incorporated. Patients receiving LRRT demonstrated significantly improved progression-free survival (3-year PFS rate, 53.2 % vs 31.2 %, p < 0.001). After PSM analysis, there were 244 patients in the LRRT group and 122 patients in the non-LRRT group. Multivariable analysis indicated that LRRT was not an independent prognostic factor in the matched cohort (HR, 1.25, 95 % CI, 0.92-1.69, p = 0.156). Subgroup analysis among the matched cohort showed a significant increase in PFS for patients with oligo metastatic disease (OMD) who received LRRT (3-year PFS rate, 70.6 % vs 49.3 %, p = 0.043). In contrast, no such benefit was observed in patients with poly metastatic disease (PMD, 3-year PFS rate, 35.8 % vs 27.8 %, p = 0.17). Furthermore, LRRT significantly enhanced survival in patients with undetectable EBV DNA2-6 cycles (3-year PFS rate, 57.9 % vs. 43.4 %, p = 0.043), whereas no survival improvement was noted in patients with detectable EBV DNA2-6 cycles (16.2 % vs. 20.3 %, p = 0.21). CONCLUSION LRRT could prolong PFS in M1-NPC patients. OMD and undetectable EBV DNA2-6 cycles are potential indicators for selecting beneficiaries from LRRT.
Collapse
Affiliation(s)
- Shui-Qing He
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China
| | - Guo-Ying Liu
- Department of Oncology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ya-Hui Yu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lin Wang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China
| | - Guo-Yi Zhang
- Department of Radiation Oncology, Cancer Center, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Ding-Sheng Peng
- Department of Radiation Oncology, Huizhou Central People's Hospital, No. 41, Eling North Road, Hui cheng District, Huizhou 516008, PR China
| | - Wei-Xin Bei
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China
| | - Chun-Lan Chen
- Department of Radiation Oncology, Cancer Center, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Shu-Hui Lv
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China
| | - Ze-Yu Zhao
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China
| | - Ying Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060 Guangzhou, China.
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 510060, Guangzhou, China.
| |
Collapse
|
3
|
Sun H, Li M, Huang W, Zhang J, Wei S, Yang Y, Wang Z, Ye S, Gong H, Zhang Y, Li J, Song H, Wang L, Chen X, Lin H, Ding G, Li H, Zheng A, Ma X, Chen S, Liu L, Zhang K, Fu C, Liu W, Wang J, Zhang X, Liu T, Han D, Zhao Q, Wu P, Yuan Q, Tian L, Zhang P, Wu X, Chen F, Zhang Z, Li B. Thoracic Radiotherapy Improves the Survival in Patients With EGFR-Mutated Oligo-Organ Metastatic Non-Small Cell Lung Cancer Treated With Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors: A Multicenter, Randomized, Controlled, Phase III Trial. J Clin Oncol 2025; 43:412-421. [PMID: 39374473 DOI: 10.1200/jco.23.02075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/05/2024] [Accepted: 08/12/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE This multicenter, randomized, phase III clinical trial (Northern Radiation Oncology Group of China-002) focused on patients with oligo-organ metastatic non-small cell lung cancer (NSCLC) who have epidermal growth factor receptor (EGFR) mutations. We aimed to investigate whether first-line concurrent thoracic radiotherapy (TRT) and EGFR-tyrosine kinase inhibitors (TKIs), compared with TKIs alone, could achieve better survival. MATERIALS AND METHODS The patients in the TKI plus TRT group received 60 Gy to primary lung tumor and positive regional lymph nodes. Radiotherapy for metastases to other sites was determined by clinicians. The primary end point was the progression-free survival (PFS). Secondary end points included overall survival (OS) and treatment-related adverse events (TRAEs). The first and second interim analyses were performed in March 2021 and March 2022. RESULTS Between April 14, 2016, and February 25, 2022, a total of 118 patients were enrolled. Compared with the TKI alone group, the TKI plus TRT group achieved significantly better PFS (hazard ratio [HR], 0.57; P = .004) and OS (HR, 0.62; P = .029). The median PFS was 10.6 months in the TKI alone group and 17.1 months in the TKI plus TRT group. The median OS was 26.2 months and 34.4 months in the TKI alone group and TKI plus TRT group, respectively. The TKI plus TRT group showed better local control but was associated with a higher incidence of severe TRAEs (11.9% v 5.1%). CONCLUSION For patients with EGFR-mutated oligo-organ metastatic NSCLC treated with first-line EGFR-TKIs, concurrent TRT improves the PFS and OS, and TRAEs are acceptable and tolerable.
Collapse
Affiliation(s)
- Hongfu Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Minghao Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Jian Zhang
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shihong Wei
- Department of Radiation Oncology, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Yongjing Yang
- Department of Radiation Oncology, Jilin Cancer Hospital, Changchun, China
| | - Zhongtang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Shucheng Ye
- Department of Radiation Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Heyi Gong
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaowen Zhang
- Department of Radiation Oncology, Anyang Tumor Hospital, the Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Henan Medical Key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, China
| | - Jie Li
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Haixia Song
- Department of Radiation Oncology, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Lifang Wang
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangming Chen
- Department of Clinical Oncology, Taian City Central Hospital, Taian, China
| | - Haiqun Lin
- Department of Oncology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gaofeng Ding
- Department of Radiation Oncolog, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Li
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Anping Zheng
- Department of Radiation Oncology, Anyang Tumor Hospital, the Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Henan Medical Key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, China
| | - Xuezhen Ma
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, China
| | - ShaoShui Chen
- Department of Radiation Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Liping Liu
- Department of Oncology, Jining No.1 People's Hosptial, Jining, China
| | - Kaixian Zhang
- Department of Radiation Oncology, Tengzhou Central People's Hospital, Tengzhou, China
| | - Chengrui Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Wenzhi Liu
- Department of Radiation Oncology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jing Wang
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xiaqin Zhang
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Tingting Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Dan Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Qian Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Peipei Wu
- Department of Oncology, Jining No.1 People's Hosptial, Jining, China
| | - Qianqian Yuan
- Department of Radiation Oncology, Tengzhou Central People's Hospital, Tengzhou, China
| | - LiJun Tian
- Department of Radiation Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Ping Zhang
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xueqin Wu
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Fei Chen
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Zicheng Zhang
- Department of Radiation Oncology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
4
|
Banerjee S, Booth CM, Bruera E, Büchler MW, Drilon A, Fry TJ, Ghobrial IM, Gianni L, Jain RK, Kroemer G, Llovet JM, Long GV, Pantel K, Pritchard-Jones K, Scher HI, Tabernero J, Weichselbaum RR, Weller M, Wu YL. Two decades of advances in clinical oncology - lessons learned and future directions. Nat Rev Clin Oncol 2024; 21:771-780. [PMID: 39354161 DOI: 10.1038/s41571-024-00945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/03/2024]
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK.
- The Institute of Cancer Research, London, UK.
| | | | - Eduardo Bruera
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer, Unit 1414, Houston, TX, USA.
| | - Markus W Büchler
- Botton-Champalimaud Pancreatic Cancer, Champalimaud Foundation, Lisbon, Portugal.
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA.
| | - Terry J Fry
- Department of Paediatrics and Immunology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Aurora, CO, USA.
| | - Irene M Ghobrial
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Harvard Medical School, Boston, MA, USA.
| | | | - Rakesh K Jain
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia.
| | - Klaus Pantel
- Institute of Tumour Biology, University Cancer Center Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Barcelona, Spain.
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA.
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Reddy JP, Sherry AD, Fellman B, Liu S, Bathala T, Haymaker C, Cohen L, Smith BD, Ramirez D, Shaitelman SF, Chun SG, Medina-Rosales M, Teshome M, Brewster A, Barcenas CH, Reuben A, Ghia AJ, Ludmir EB, Weed D, Shah SJ, Mitchell MP, Woodward WA, Gomez DR, Tang C. Adding Metastasis-Directed Therapy to Standard-of-Care Systemic Therapy for Oligometastatic Breast Cancer (EXTEND): A Multicenter, Randomized Phase 2 Trial. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03522-3. [PMID: 39486645 DOI: 10.1016/j.ijrobp.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
PURPOSE Prior evidence suggests a progression-free survival (PFS) benefit from adding metastasis-directed therapy (MDT) to standard-of-care (SOC) systemic therapy for patients with some oligometastatic solid tumors. Randomized trials testing this hypothesis in breast cancer have yet to be published. We sought to determine whether adding MDT to SOC systemic therapy improves PFS in oligometastatic breast cancer. METHODS AND MATERIALS External Beam Radiation to Eliminate Nominal Metastatic Disease is a multicenter phase 2 randomized basket trial testing the addition of MDT to SOC systemic therapy in patients with ≤5 metastases (NCT03599765). Patients were randomly assigned 1:1 to MDT (definitive local treatment to all sites of disease, plus SOC systemic therapy) or to SOC systemic therapy-only. Primary endpoint was PFS, and secondary endpoints included overall survival, time to subsequent line of systemic therapy, and time to the appearance of new metastases. Exploratory analyses included quality of life and systemic immune response measures. RESULTS From September 2018 through July 2022, 22 and 21 patients were randomly assigned to the MDT and no-MDT arms, respectively. At a median follow-up of 24.8 months, PFS was not improved with the addition of MDT to SOC systemic therapy (median PFS 15.6 months MDT vs 24.9 months no-MDT [hazard ratio, 0.91; 95% CI, 0.34-2.48; P = .86]). Similarly, MDT did not improve overall survival, time to subsequent line of systemic therapy, or time to the appearance of new metastases (all P > .05). No significant differences were found in quality of life measures, systemic T-cell activation, or T-cell stimulatory cytokine concentration. CONCLUSIONS Among patients with oligometastatic breast cancer, the addition of MDT to SOC systemic therapy did not improve PFS. These findings suggest that MDT may have no systemic benefit in otherwise unselected patients with oligometastatic breast cancer, although this trial was limited by a heterogeneous and small sample size and overperformance of both treatment arms.
Collapse
Affiliation(s)
- Jay P Reddy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Alexander D Sherry
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tharakeswara Bathala
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lorenzo Cohen
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin D Smith
- Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Ramirez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simona F Shaitelman
- Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen G Chun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Medina-Rosales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mediget Teshome
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abenaa Brewster
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos H Barcenas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Thoracic-Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amol J Ghia
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Weed
- Community Physician Network, Radiation Oncology Care, Indianapolis, Indiana; The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shalin J Shah
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa P Mitchell
- Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendy A Woodward
- Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad Tang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Genitourinary Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
6
|
Jiang Y, Cao H, Deng H, Guan L, Langthasa J, Colburg DRC, Melemenidis S, Cotton RM, Aleman J, Wang XJ, Graves EE, Kalbasi A, Pu K, Rao J, Le QT. Gold-siRNA supraclusters enhance the anti-tumor immune response of stereotactic ablative radiotherapy at primary and metastatic tumors. Nat Biotechnol 2024:10.1038/s41587-024-02448-0. [PMID: 39448881 DOI: 10.1038/s41587-024-02448-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Strategies to enhance the anti-tumor immune response of stereotactic ablative radiotherapy (SABR) at primary tumors and abscopal sites are under intensive investigation. Here we report a metabolizable binary supracluster (BSCgal) that combines gold nanoclusters as radiosensitizing adjuvants with small interfering RNA (siRNA) targeting the immunosuppressive mediator galectin-1 (Gal-1). BSCgal comprises reversibly crosslinked cationic gold nanoclusters and siRNA complexes in a polymer matrix that biodegrades over weeks, facilitating clearance (90.3% in vivo clearance at 4 weeks) to reduce toxicity. The particle size well above the renal filtration threshold facilitates passive delivery to tumors. Using mouse models of head and neck cancer, we show that BSCgal augments the radiodynamic and immunotherapeutic effects of SABR at the primary and metastatic tumors by promoting tumor-inhibitory leukocytes, upregulating cytotoxic granzyme B and reducing immunosuppressive cell populations. It outperforms SABR plus Gal-1 antagonists, chemoradiation drug cisplatin or PD-1 inhibitor. This work presents a translatable strategy to converge focal radiosensitization with targeted immune checkpoint silencing for personalized radioimmunotherapy.
Collapse
Affiliation(s)
- Yuyan Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Hongbin Cao
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Huaping Deng
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Li Guan
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Jimpi Langthasa
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | | | - Renee M Cotton
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - John Aleman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Xiao-Jing Wang
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jianghong Rao
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
8
|
Christ SM, Breitenstein M, Heesen P, Turner B, Muehlematter UJ, Pohl K, Willmann J, Maurer A, Nagpal SK, Ahmadsei M, Badra Vlaskou E, Looman EL, Heusel AE, Mayinger M, Balermpas P, Wicki A, Andratschke N, Balboni T, Anh Huynh M, Huellner M, Guckenberger M. Clinical management of oligometastatic cancer: Applying multidisciplinary tumor board recommendations in practice. Clin Transl Radiat Oncol 2024; 48:100838. [PMID: 39224662 PMCID: PMC11367634 DOI: 10.1016/j.ctro.2024.100838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Aims Multidisciplinary tumor boards (MDTs) are an integral part of ensuring high-quality, evidence-based and personalized cancer care. In this study, we aimed to evaluate the adherence to and implementation of MDT recommendations in patients with oligometastatic disease (OMD). Methods We screened all oncologic positron emission tomography (PET) scans conducted at a single comprehensive cancer center in 2020. Patients were included if they had evidence of imaging-based OMD from a solid organ malignancy on the index scans, had their OMD case discussed at an MDT, and were treated and followed up at the same center. A switch away from the MDT-recommended treatment modalities was classified as a major deviation; non-MDT-mandated adjustments to a recommended treatment modality were coded as minor deviation. Clinical data was obtained via chart review; statistical calculations were computed using the R software. Results After review of PET and/or concurrent brain scans, 787 cases of OMD were identified. Thereof, 347 (44.1 %) cases were discussed at MDT, of which 331 (42.1 %) were therapeutically managed and subsequently followed. The three most commonly recommended therapies were systemic therapy (35.6 %), multimodality treatment including definitive local therapy (17.8 %), and radiotherapy (13.9 %). A major deviation was recorded in 16.3 % of cases (most commonly: none of the MDT-recommended treatment modalities were performed: 19 (35.2 %); not all MDT-planned treatment modalities were performed: 12 (22.2 %); and additional treatment modality was performed: 11 (20.3 %). A minor deviation was found in 1.5 % of cases. On multivariable regression, number of distant metastases (n > 1) was associated with a major deviation (OR: 1.85; 95 % CI, 1.0-3.52). Major deviations were associated with a significantly worse OS (p = 0.0034). Conclusions Adherence to and implementation of MDT recommendations in OMD patients was generally high (83.7%). Major deviations might be further reduced by more careful and elaborate discussions of OMD patient characteristics s and patient preferences.
Collapse
Affiliation(s)
- Sebastian M. Christ
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | | | - Philip Heesen
- Faculty of Medicine, University of Zurich, Zurich, CH, Switzerland
| | - Brandon Turner
- Dept. of Radiation Oncology, Brigham and Women’s Hospital, Dana Farber Cancer Institute, Boston, MA, USA
| | - Urs J. Muehlematter
- Dept. of Nuclear Medicine, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Kaspar Pohl
- Faculty of Medicine, University of Zurich, Zurich, CH, Switzerland
| | - Jonas Willmann
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Alexander Maurer
- Dept. of Nuclear Medicine, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | | | - Maiwand Ahmadsei
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Eugenia Badra Vlaskou
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Esmée L. Looman
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Astrid E. Heusel
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Michael Mayinger
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Panagiotis Balermpas
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Andreas Wicki
- Dept. of Medical Oncology & Hematology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Nicolaus Andratschke
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Tracy Balboni
- Dept. of Radiation Oncology, Brigham and Women’s Hospital, Dana Farber Cancer Institute, Boston, MA, USA
| | - Mai Anh Huynh
- Dept. of Radiation Oncology, Brigham and Women’s Hospital, Dana Farber Cancer Institute, Boston, MA, USA
| | - Martin Huellner
- Dept. of Nuclear Medicine, University Hospital & University of Zurich, Zurich, CH, Switzerland
| | - Matthias Guckenberger
- Dept. of Radiation Oncology, University Hospital & University of Zurich, Zurich, CH, Switzerland
| |
Collapse
|
9
|
Zuur LG, de Barros HA, van Oosterom MN, Berrens AC, Donswijk ML, Hendrikx JJMA, Bekers EM, Vis AN, Wit EM, van Leeuwen FB, van der Poel HG, van Leeuwen PJ. 99mTcPSMA-radioguided surgery in oligorecurrent prostate cancer: the randomised TRACE-II trial. BJU Int 2024; 134:81-88. [PMID: 38346924 DOI: 10.1111/bju.16297] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
OBJECTIVE To investigate whether combination treatment of prostate-specific membrane antigen (PSMA)-based radioguided surgery (RGS) with short-term androgen deprivation therapy (ADT) improves oncological outcomes in men with oligorecurrent prostate cancer (PCa) as compared to treatment with short-term ADT only. METHODS The TRACE-II study is an investigator-initiated, prospective, randomised controlled clinical trial. Patients (aged >18 years) with hormone-sensitive recurrent PCa after radical prostatectomy or radiotherapy (brachytherapy or external beam radiotherapy), with involvement of ≤2 lymph nodes or local oligorecurrent disease within the pelvis as determined by PSMA positron emission tomography (PET)/computed tomography (CT) are randomly assigned in a 1:1 ratio between 6-month ADT (Arm A) or 6-month ADT plus RGS (Arm B). The primary objective is to determine clinical progression-free survival (CPFS) at 24 months. After PSMA-RGS, CPFS is defined as the time between the start of treatment and the appearance of a re-recurrence (any N1 or M1) as suggested by PSMA-PET/CT or symptoms related to progressive PCa, or death from any cause. The secondary objectives include metastasis-free survival at 2, 5 and 10 years, biochemical progression-free survival at 2 years, and patient-reported quality of life at 2, 5 and 10 years. A total of 60 patients, 30 per arm, will be included. The trial is powered (80%) to detect at least a 30% absolute difference in CPFS between the two study arms in the period 2 years after randomisation. We expect to enrol the required participants in 3 years. The study has an expected duration of 5 years in total. CONCLUSIONS Combining RGS with short-term ADT might be oncologically beneficial for patients with oligorecurrent PCa. In this first randomised controlled trial, we are investigating the potential oncological benefits of this combined treatment, while also focusing on maintaining quality of life.
Collapse
Affiliation(s)
- Lotte G Zuur
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
| | - Hilda A de Barros
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
| | - Matthias N van Oosterom
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anne-Claire Berrens
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maarten L Donswijk
- Department of Nuclear Medicine, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Nuclear Medicine, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Clinical Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Elise M Bekers
- Department of Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - André N Vis
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Esther M Wit
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
| | - Fijs B van Leeuwen
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Centre, Leiden, The Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Netherlands Prostate Cancer Network, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Wang B, Pan J, Zhang T, Ni X, Wei Y, Li X, Fang B, Hu X, Gan H, Wu J, Wang H, Ye D, Zhu Y. Protocol for CHAMPION study: a prospective study of maximal-cytoreductive therapies for patients with de novo metastatic hormone-sensitive prostate cancer who achieve oligopersistent metastases during systemic treatment with apalutamide plus androgen deprivation therapy. BMC Cancer 2024; 24:643. [PMID: 38796422 PMCID: PMC11127327 DOI: 10.1186/s12885-024-12395-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/16/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND The proposed trial is to examine the feasibility of prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT)-guided cytoreduction plus apalutamide and androgen deprivation therapy (ADT) for newly diagnosed metastatic hormone-sensitive prostate cancer (mHSPC) at oligometastatic state. METHODS CHAMPION (NCT05717582) is an open-label, single-arm, phase II trial, planning to enroll newly diagnosed mHSPC cases with oligometastases (≤ 10 distant metastatic sites in conventional imaging). Patients will receive 6 cycles of apalutamide plus ADT. Patients with oligometastatic disease at PSMA PET/CT after 3 treatment cycles will receive cytoreductive radical prostatectomy. PSMA PET/CT-guided metastasis-directed external radiation therapy will be determined by the investigators. Apalutamide plus ADT will be continued for 2 weeks postoperatively. The primary endpoint is the proportion of patients with undetectable prostate-specific antigen (PSA), no disease progression, and no symptom deterioration after 6 cycles of apalutamide plus ADT. Secondary endpoints include the percentage of patients with PSA ≤ 0.2 ng/mL and oligometastases by the end of 3 treatment cycles, PSA response rate, and safety. Fleming's two-stage group sequential design will be adopted in the study, where the null hypothesis is that the rate of patients with an undetectable PSA is ≤ 40% after 6 cycles of treatment, while the alternate hypothesis is an undetectable PSA of > 60%; with one-sided α = 0.05, power = 0.80, and an assumed dropout rate of 10%, the required number of patients for an effective analysis is 47. Enrolment in the study commenced in May 2023. DISCUSSION The multi-modal therapy based on treatment response may improve the prognosis of newly diagnosed mHSPC patients with oligometastases. TRIAL REGISTRATION The study is registered with Clinical Trials.Gov (NCT05717582). Registered on 8th February 2023.
Collapse
Affiliation(s)
- Beihe Wang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoxin Hu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongkai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Huguet F, Riou O, Pasquier D, Modesto A, Quéro L, Michalet M, Bordron A, Schipman B, Orthuon A, Lisbona A, Vendrely V, Jaksic N. Radiation therapy of the primary tumour and/or metastases of digestive metastatic cancers. Cancer Radiother 2024; 28:66-74. [PMID: 37806823 DOI: 10.1016/j.canrad.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 10/10/2023]
Abstract
Metastatic gastrointestinal cancer is not an uncommon situation, especially for pancreatic, gastric, and colorectal cancers. In this setting, few data are available on the impact of the treatment of the primary tumour. Oligometastatic disease is associated with longer survival in comparison with more advanced disease. Metastasis-directed therapy, such as stereotactic body radiotherapy, seems related to better outcomes, but the level of evidence is low. In most tumour locations, prospective data are very scarce and inclusion in ongoing trials is strongly recommended.
Collapse
Affiliation(s)
- F Huguet
- Service d'oncologie radiothérapie, hôpital Tenon, AP-HP, DMU Orphé, Sorbonne université, Paris, France; Laboratory of Cancer Biology and Therapeutics, centre de recherche Saint-Antoine, U938, Inserm, Paris, France.
| | - O Riou
- Institut de recherche en cancérologie de Montpellier, U1194, Inserm, université de Montpellier, Montpellier, France; Fédération universitaire d'oncologie radiothérapie d'Occitanie Méditerranée, ICM, institut régional du cancer de Montpellier, Montpellier, France
| | - D Pasquier
- Service d'oncologie radiothérapie, centre Oscar-Lambret, Lille, France; Université de Lille, CNRS, école centrale de Lille, UMR 9189 - CRIStAL, Lille, France
| | - A Modesto
- Département de radiothérapie, institut universitaire du cancer de Toulouse, Toulouse, France; Centre de recherche du cancer de Toulouse, UMR 1037, Inserm, université Toulouse-III Paul-Sabatier, Toulouse, France
| | - L Quéro
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, AP-HP Nord, DMU Icare, Paris, France; Université Paris Cité, U1160, Inserm, Paris, France
| | - M Michalet
- Institut de recherche en cancérologie de Montpellier, U1194, Inserm, université de Montpellier, Montpellier, France; Fédération universitaire d'oncologie radiothérapie d'Occitanie Méditerranée, ICM, institut régional du cancer de Montpellier, Montpellier, France
| | - A Bordron
- Département de radiothérapie, centre hospitalier universitaire de Brest, Brest, France
| | - B Schipman
- Institut de cancérologie de Bourgogne, Dijon, France
| | - A Orthuon
- Institut de cancérologie de Bourgogne, Dijon, France
| | - A Lisbona
- Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France
| | - V Vendrely
- Service d'oncologie radiothérapie, hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - N Jaksic
- Institut de cancérologie et radiothérapie Brétillien, Saint-Malo, France
| |
Collapse
|
12
|
García-Figueiras R, Baleato-González S, Luna A, Padhani AR, Vilanova JC, Carballo-Castro AM, Oleaga-Zufiria L, Vallejo-Casas JA, Marhuenda A, Gómez-Caamaño A. How Imaging Advances Are Defining the Future of Precision Radiation Therapy. Radiographics 2024; 44:e230152. [PMID: 38206833 DOI: 10.1148/rg.230152] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Radiation therapy is fundamental in the treatment of cancer. Imaging has always played a central role in radiation oncology. Integrating imaging technology into irradiation devices has increased the precision and accuracy of dose delivery and decreased the toxic effects of the treatment. Although CT has become the standard imaging modality in radiation therapy, the development of recently introduced next-generation imaging techniques has improved diagnostic and therapeutic decision making in radiation oncology. Functional and molecular imaging techniques, as well as other advanced imaging modalities such as SPECT, yield information about the anatomic and biologic characteristics of tumors for the radiation therapy workflow. In clinical practice, they can be useful for characterizing tumor phenotypes, delineating volumes, planning treatment, determining patients' prognoses, predicting toxic effects, assessing responses to therapy, and detecting tumor relapse. Next-generation imaging can enable personalization of radiation therapy based on a greater understanding of tumor biologic factors. It can be used to map tumor characteristics, such as metabolic pathways, vascularity, cellular proliferation, and hypoxia, that are known to define tumor phenotype. It can also be used to consider tumor heterogeneity by highlighting areas at risk for radiation resistance for focused biologic dose escalation, which can impact the radiation planning process and patient outcomes. The authors review the possible contributions of next-generation imaging to the treatment of patients undergoing radiation therapy. In addition, the possible roles of radio(geno)mics in radiation therapy, the limitations of these techniques, and hurdles in introducing them into clinical practice are discussed. ©RSNA, 2024 Test Your Knowledge questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Roberto García-Figueiras
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Sandra Baleato-González
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Antonio Luna
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Anwar R Padhani
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Joan C Vilanova
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Ana M Carballo-Castro
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Laura Oleaga-Zufiria
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Juan Antonio Vallejo-Casas
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Ana Marhuenda
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| | - Antonio Gómez-Caamaño
- From the Department of Radiology, Division of Oncologic Imaging (R.G.F., S.B.G.), and Department of Radiation Oncology (A.M.C.C., A.G.C.), Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706 Santiago de Compostela, Spain; Department of Advanced Medical Imaging, Grupo Health Time, Sercosa (Servicio Radiologia Computerizada, Clínica Las Nieves, Jaén, Spain (A.L.); Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England (A.R.P.); Department of Radiology, Clínica Girona and Hospital Santa Caterina, Girona, Spain (J.C.V.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.Z.); Unidad de Gestión Clínica de Medicina Nuclear, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Córdoba, Spain (J.A.V.C.); and Department of Radiology, Instituto Valenciano de Oncología, Valencia, Spain (A.M.)
| |
Collapse
|
13
|
Gutman MJ, Serra LM, Koshy M, Katipally RR. SBRT for Liver Tumors: What the Interventional Radiologist Needs to Know. Semin Intervent Radiol 2024; 41:1-10. [PMID: 38495259 PMCID: PMC10940045 DOI: 10.1055/s-0043-1778657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
This review summarizes the clinical evidence supporting the utilization of stereotactic body radiotherapy (SBRT) for liver tumors, including hepatocellular carcinoma, liver metastases, and cholangiocarcinoma. Emerging prospective evidence has demonstrated the benefit and low rates of toxicity across a broad range of clinical contexts. We provide an introduction for the interventional radiologist, with a discussion of underlying themes such as tumor dose-response, mitigation of liver toxicity, and the technical considerations relevant to performing liver SBRT. Ultimately, we recommend that SBRT should be routinely included in the armamentarium of locoregional therapies for liver malignancies, alongside those liver-directed therapies offered by interventional radiology.
Collapse
Affiliation(s)
- Michael J. Gutman
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Lucas M. Serra
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Matthew Koshy
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Rohan R. Katipally
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
14
|
Llacer-Moscardo C, Moureau-Zabotto L, Ollivier L, Helfré S, Ducassou A, Bonvalot S, Sunyach MP, Sargos P, Gillon P, Firmin N, Le Péchoux C, Thariat J. Management of oligometastatic/metastatic sarcomas and place of local treatments with focus on modern radiotherapy approaches. Cancer Radiother 2024; 28:93-102. [PMID: 38212215 DOI: 10.1016/j.canrad.2023.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/09/2023] [Accepted: 06/29/2023] [Indexed: 01/13/2024]
Abstract
Soft tissue sarcomas are a rare and heterogeneous disease. For localized disease, treatment is based on surgery and radiotherapy with or without chemotherapy depending on risk factors. Upfront metastases are present in 7 to 20% of cases, and are localized to the lungs in most of cases. Disseminated disease is generally considered incurable but in selected cases, aggressive local treatment of metastases allowed long survival. Treatment of primary tumour is often debated. Our purpose is to evaluate the literature concerning the role of radiotherapy in the management of primary metastatic soft tissue sarcomas.
Collapse
Affiliation(s)
- C Llacer-Moscardo
- Radiation oncology department, institut du cancer de Montpellier (ICM), 208, avenue des Apothicaires, parc Euromédecine, 34298 Montpellier cedex 5, France.
| | - L Moureau-Zabotto
- Department of radiotherapy, centre de radiothérapie du Pays d'Aix, avenue Henri-Pontier, 13100 Aix-en-Provence, France
| | - L Ollivier
- Department of radiotherapy, institut de cancérologie de l'Ouest (ICO), centre René-Gauducheau, Saint-Herblain, France
| | - S Helfré
- Department of radiotherapy, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - A Ducassou
- Department of radiotherapy, IUCT Oncopole, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France
| | - S Bonvalot
- Department of oncological surgery, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - M-P Sunyach
- Department of radiotherapy, centre Léon-Bérard, 28, promenade Léa-et-Napoléon-Bullukian, 69008 Lyon, France
| | - P Sargos
- Department of radiotherapy, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France
| | - P Gillon
- Department of radiotherapy, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France
| | - N Firmin
- Radiation oncology department, institut du cancer de Montpellier (ICM), 208, avenue des Apothicaires, parc Euromédecine, 34298 Montpellier cedex 5, France
| | - C Le Péchoux
- Department of radiotherapy, Gustave-Roussy, 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | - J Thariat
- Department of radiotherapy, centre François-Baclesse, 12, rue Jean-Baptiste-Colbert, 14000 Caen, France
| |
Collapse
|
15
|
Wang L, Katipally RR, Liang HL, Yang K, Pitroda SP, He C, Weichselbaum RR. RNA m 6A methylation and MDSCs: Roles and therapeutic implications for radiotherapy. MED 2023; 4:863-874. [PMID: 38070481 PMCID: PMC10751059 DOI: 10.1016/j.medj.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/29/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023]
Abstract
Emerging evidence suggests that local tumor radiotherapy reshapes the repertoire of circulating myeloid-derived suppressor cells (MDSCs) and leads to their infiltration into the tumor microenvironment, which poses a major obstacle for radiotherapy efficacy. Recent findings have identified RNA m6A modification at the nexus of both anti-tumor immunity and radiation response. Here, we examine the mechanisms by which this RNA modification modulates the immune milieu of the radiation-remodeled tumor microenvironment. We discuss potential therapeutic interventions targeting m6A machinery to improve radiotherapy response.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| | - Rohan R Katipally
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Sean P Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
16
|
Jia Q, Zhang R, Yan H, Feng Y, Sun F, Yang Z, Qiao C, Mou X, Tian J, Wang Z. An Activatable Near-Infrared Fluorescent Probe for Precise Detection of the Pulmonary Metastatic Tumors: A Traditional Molecule Having a Stunning Turn. Angew Chem Int Ed Engl 2023; 62:e202313420. [PMID: 37779105 DOI: 10.1002/anie.202313420] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/03/2023]
Abstract
An accurate detection of lung metastasis is of great significance for making better treatment choices and improving cancer prognosis, but remains a big challenge in clinical practice. In this study, we propose a reinventing strategy to develop a pH-activatable near-infrared (NIR) fluorescent nanoprobe, pulmonary metastasis tracer (denoted as PMT), based on assembly of NIR dye IR780 and calcium phosphate (CaP). By delicately tuning the intermolecular interactions during the assembly process and dye doping content, as well as the synthetic condition of probe, the fluorescence of PMT could be finely adjusted via the tumor acidity-triggered disassembly. Notably, the selected PMT9 could sharply convert subtle pH variations into a distinct fluorescence signal to generate high fluorescence ON/OFF contrast, dramatically reducing the background signals. Benefiting from such preferable features, PMT9 is able to precisely identify not only the tumor sites in orthotopic lung cancer models but also the pulmonary metastases in mice with remarkable signal-to-background ratio (SBR). This study provides a unique strategy to turn shortcomings of traditional dye IR780 during in vivo imaging into advantages and further expand the application of fluorescent probe to image lung associated tumor lesions.
Collapse
Affiliation(s)
- Qian Jia
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Ruili Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Haohao Yan
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Yanbin Feng
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Fang Sun
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zuo Yang
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Chaoqiang Qiao
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Xiaocheng Mou
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Jie Tian
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM) Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| |
Collapse
|
17
|
Liao X, Kishi K, Du K, Komaki R, Mizoe J, Aikawa G, Zheng W, Pan C. Risk factors of local control in adrenal metastases treated by stereotactic body radiation therapy - a systematic review and meta-analysis. Front Oncol 2023; 13:1193574. [PMID: 38045003 PMCID: PMC10691549 DOI: 10.3389/fonc.2023.1193574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 10/19/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose This study is aimed to explore risk factors affect the therapy outcomes of adrenal metastases (AM) for stereotactic body radiation therapy (SBRT) and guide clinical dose selection. Methods and materials PubMed, Embase and Web of Science were searched in September 22, 2022 in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines (PRISMA). Subgroup analysis and meta-regression were used to search for sources of heterogeneity and identify risky outcomes factors. Publication bias test and sensitivity analysis were also conducted. Results Thirty-three studies with full text from 2009 to 2022 about AM with SBRT on 1483 patients were included. Pooled 1- and 2-year local control (LC) and overall survival(OS) were 81.7% (95% confidence interval [CI], 75.6%-86.5%), 62.8% (95% CI, 53.8%-71.8%), 67.4% (95%CI, 61.8%-73.1%) and 46.5% (95%CI, 40.4%-52.6%), respectively. Biological effective dose (BED, α/β=10Gy) and dose per fraction affected 1-year LC (Qm=23.89, 15.10; P<0.0001, 0.0001). In the range of 60-80Gy (BED10), the group of dose per fraction ≥ 9Gy achieved the excellent 1-year LC (< 9Gy: ≥ 9Gy =78%, 91%; χ2 = 10.16, P = 0.001). Tracking technology significantly affected 1- and 2-year OS (Qm = 5.73, 8.75; P = 0.017, 0.003) and high tracking adoption group showed excellent 1- and 2- year OS (78.7% [95%CI, 68.6%- 88.9%]; and 62.9% [95%CI, 53.1%-72.7%]). Conclusion Increasing the dose per fraction appropriately may help control locally AM lesious. Tracking technology might contribute to improve survival of advanced patients with AM. But these results need prospective studies to verify them.
Collapse
Affiliation(s)
- Xuehong Liao
- Department of Pathology, Zhongshan Hospital, Xiamen University, Xiamen, China
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Kazushi Kishi
- Department of Radiation Oncology, National Disaster Medical Center, National Hospital Organization (NHO), Incorporated Administrative Agency 3256 Tachitawa City, Tokyo, Japan
| | - Kaixin Du
- Department of Radiation Oncology, Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ritsuko Komaki
- Department of Radiation Oncology, Emeritus of The University of Texas M.D. Anderson Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Junetsu Mizoe
- Department of Sapporo High Functioning Radiotherapy Center, Hokkaido Ohno Memorial Hospital, Sapporo, Japan
| | - Gosuke Aikawa
- Department of Sapporo High Functioning Radiotherapy Center, Hokkaido Ohno Memorial Hospital, Sapporo, Japan
| | - Wei Zheng
- Department of Pathology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Chao Pan
- Department of Pathology, Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
18
|
Zheng Y, Cong C, Wang Z, Liu Y, Zhang M, Zhou H, Su C, Sun M. Decreased risk of radiation pneumonitis with concurrent use of renin-angiotensin system inhibitors in thoracic radiation therapy of lung cancer. Front Med (Lausanne) 2023; 10:1255786. [PMID: 37901395 PMCID: PMC10602779 DOI: 10.3389/fmed.2023.1255786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Background Radiation pneumonitis (RP) is the primary dose-limiting toxicity associated with radiotherapy. This study aimed to observe the effects of renin-angiotensin system inhibitors in Chinese patients with lung cancer who received thoracic radiation. Methods Patients with lung cancer who received thoracic radiation at a total dose of ≥45 Gray between October 2017 and December 2022 were enrolled in this study. We retrospectively evaluated the factors influencing grade 2 or higher RP. Results A total of 320 patients were enrolled in this study; 62 patients were identified as angiotensin receptor blockers or angiotensin-converting enzyme inhibitor users. Additionally, 99 patients (30.9%) had grade 2 or higher RP, and the incidence in the renin-angiotensin system inhibitor group was 17.7% (11 out of 62 patients). Patients in the renin-angiotensin system inhibitors (RASi) group were older and had a higher percentage of males, lower percentage of ECOG score 0, higher percentage of hypertension, and higher percentage of adenocarcinoma than those in the non-RASi group. ECOG score [hazard ratio (HR) = 1.69, p = 0.009], history of smoking (HR = 1.76, p = 0.049), mean dose (HR = 3.63, p = 0.01), and RASi (HR = 0.3, p = 0.003) were independent predictive factors for RP. All subgroups benefited from RASi. Conclusion This study showed that oral RASi administration has the potential to mitigate the incidence of grade 2 or higher RP in patients with lung cancer undergoing thoracic radiotherapy. To validate and further substantiate these findings, additional prospective research is warranted.
Collapse
Affiliation(s)
- Yawen Zheng
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
| | - Changsheng Cong
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
| | - Zewen Wang
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
| | - Yanan Liu
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Mingyan Zhang
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Hao Zhou
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
| | - Chen Su
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
| | - Meili Sun
- Department of Oncology, Central Hospital Affiliated To Shandong First Medical University, Jinan, China
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, China
| |
Collapse
|
19
|
Piombino C, Oltrecolli M, Tonni E, Pirola M, Matranga R, Baldessari C, Pipitone S, Dominici M, Sabbatini R, Vitale MG. De Novo Metastatic Prostate Cancer: Are We Moving toward a Personalized Treatment? Cancers (Basel) 2023; 15:4945. [PMID: 37894312 PMCID: PMC10605467 DOI: 10.3390/cancers15204945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
De novo metastatic hormone-sensitive PC (mHSPC) accounts for 5-10% of all prostate cancer (PC) diagnoses but it is responsible for nearly 50% of PC-related deaths. Since 2015, the prognosis of mHSPC has slightly improved thanks to the introduction of new hormonal agents and chemotherapy combined with androgen deprivation therapy from the first-line setting. This review describes the current therapeutic opportunities for de novo mHSPC, focusing on potential molecular biomarkers identified in the main clinical trials that have modified the standard of care, the genomic features of de novo mHSPC, and the principal ongoing trials that are investigating new therapeutic approaches and the efficacy of a biomarker-guided treatment in this setting. The road toward personalized treatment for de novo mHSPC is still long, considering that the randomized clinical trials, which have furnished the basis of the current therapeutic options, stratified patients according to clinical criteria that did not necessarily reflect the biological rationale of the chosen therapy. The role of transcriptomic profiling of mHSPC as a predictive biomarker requires further validation, and it remains to be ascertained how the genomic variants detected in mHSPC, which are regarded as predictive in the castration-resistant disease, can be exploited in the mHSPC setting.
Collapse
Affiliation(s)
- Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Cinza Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| |
Collapse
|
20
|
Chen Y, Yu R, Liu Y. Combine radiotherapy and immunotherapy in esophageal squamous cell carcinoma. Crit Rev Oncol Hematol 2023; 190:104115. [PMID: 37633347 DOI: 10.1016/j.critrevonc.2023.104115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023] Open
Abstract
Immune checkpoint inhibitors(ICIs) have improved the survival of advanced esophageal squamous cell carcinoma (ESCC) patients. Radiotherapy is one of the common therapies to treat esophageal cancer. However, whether combination radiation therapy can increase the efficacy of immunotherapy is still up for debate. Radiotherapy combined with immunotherapy has proven to be a reliable and effective treatment for tumors, and it can work in combination with immunotherapy to achieve better anti-tumor effects. This review aims to discuss the efficacy and safety of combining radiotherapy and immunotherapy to treat ESCC by stages as well as the optimum radiotherapy dose and target volume, with a summary of clinical trials in ESCC.
Collapse
Affiliation(s)
- Yicong Chen
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruixuan Yu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongmei Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
21
|
Hu YJ, Lu TZ, Zhang H, Fang M, Chen BJ, Guo QJ, Lin SJ, Feng P, Wang Y, Jiang TC, Gong XC, Pan JJ, Li JG, Xia YF. Locoregional radiotherapy improves survival outcomes in de novo metastatic nasopharyngeal carcinoma treated with chemoimmunotherapy. ESMO Open 2023; 8:101629. [PMID: 37660406 PMCID: PMC10594020 DOI: 10.1016/j.esmoop.2023.101629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND We aimed to investigate the efficacy of locoregional radiotherapy (LRRT) in patients with de novo metastatic nasopharyngeal carcinoma (dmNPC) receiving chemotherapy combined with anti-programmed cell death receptor-1 monoclonal antibodies (anti-PD-1 mAbs) as first-line treatment and identify optimal candidates for LRRT. MATERIALS AND METHODS We enrolled patients with dmNPC receiving platinum-based palliative chemotherapy and anti-PD-1 mAbs followed or not followed by LRRT from four centers. The endpoints were progression-free survival (PFS), objective response rate (ORR), and overall survival (OS). We used the inverse probability of treatment weighting (IPTW) to balance the baseline characteristics of the LRRT and non-LRRT groups to minimize selection bias before comparative analyses. Multivariate analyses were carried out using the Cox proportional hazards model. RESULTS We included 163 patients with dmNPC (median follow-up: 22 months). The median PFS was 20 months, and the ORR was 92.0%; the median OS was not achieved. After IPTW adjustments, patients who received LRRT had a significant survival benefit over those not receiving LRRT (median PFS: 28 versus 15 months, P < 0.001). The Epstein-Barr virus DNA (EBV DNA) level after four to six cycles of anti-PD-1 mAbs [weighted hazard ratio (HR): 2.19, 95% confidence interval (CI) 1.22-3.92, P = 0.008] and LRRT (weighted HR: 0.58, 95% CI 0.34-0.99, P = 0.04) were independent prognostic factors. Patients with undetectable EBV DNA levels after four to six cycles of anti-PD-1 mAbs (early EBV DNA clearance) benefitted from LRRT (HR: 0.41, 95% CI 0.22-0.79, P = 0.008), whereas those with detectable levels did not (HR: 1.30, 95% CI 0.59-2.87, P = 0.51). CONCLUSIONS Palliative chemotherapy combined with anti-PD-1 mAbs followed by LRRT was associated with improved PFS in patients with dmNPC, especially for patients with early EBV DNA clearance.
Collapse
Affiliation(s)
- Y-J Hu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou
| | - T-Z Lu
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma (Jiangxi Cancer Hospital of Nanchang University), Nanchang; Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang
| | - H Zhang
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - M Fang
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma (Jiangxi Cancer Hospital of Nanchang University), Nanchang
| | - B-J Chen
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou
| | - Q-J Guo
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou
| | - S-J Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou
| | - P Feng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou
| | - Y Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou
| | - T-C Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou
| | - X-C Gong
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma (Jiangxi Cancer Hospital of Nanchang University), Nanchang; Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang
| | - J-J Pan
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou.
| | - J-G Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma (Jiangxi Cancer Hospital of Nanchang University), Nanchang; Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang.
| | - Y-F Xia
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou.
| |
Collapse
|
22
|
Iyengar P, All S, Berry MF, Boike TP, Bradfield L, Dingemans AMC, Feldman J, Gomez DR, Hesketh PJ, Jabbour SK, Jeter M, Josipovic M, Lievens Y, McDonald F, Perez BA, Ricardi U, Ruffini E, De Ruysscher D, Saeed H, Schneider BJ, Senan S, Widder J, Guckenberger M. Treatment of Oligometastatic Non-Small Cell Lung Cancer: An ASTRO/ESTRO Clinical Practice Guideline. Pract Radiat Oncol 2023; 13:393-412. [PMID: 37294262 DOI: 10.1016/j.prro.2023.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/07/2023] [Indexed: 06/10/2023]
Abstract
PURPOSE This joint guideline by American Society for Radiation Oncology (ASTRO) and the European Society for Radiotherapy and Oncology (ESTRO) was initiated to review evidence and provide recommendations regarding the use of local therapy in the management of extracranial oligometastatic non-small cell lung cancer (NSCLC). Local therapy is defined as the comprehensive treatment of all known cancer-primary tumor, regional nodal metastases, and metastases-with definitive intent. METHODS ASTRO and ESTRO convened a task force to address 5 key questions focused on the use of local (radiation, surgery, other ablative methods) and systemic therapy in the management of oligometastatic NSCLC. The questions address clinical scenarios for using local therapy, sequencing and timing when integrating local with systemic therapies, radiation techniques critical for oligometastatic disease targeting and treatment delivery, and the role of local therapy for oligoprogression or recurrent disease. Recommendations were based on a systematic literature review and created using ASTRO guidelines methodology. RESULTS Based on the lack of significant randomized phase 3 trials, a patient-centered, multidisciplinary approach was strongly recommended for all decision-making regarding potential treatment. Integration of definitive local therapy was only relevant if technically feasible and clinically safe to all disease sites, defined as 5 or fewer distinct sites. Conditional recommendations were given for definitive local therapies in synchronous, metachronous, oligopersistent, and oligoprogressive conditions for extracranial disease. Radiation and surgery were the only primary definitive local therapy modalities recommended for use in the management of patients with oligometastatic disease, with indications provided for choosing one over the other. Sequencing recommendations were provided for systemic and local therapy integration. Finally, multiple recommendations were provided for the optimal technical use of hypofractionated radiation or stereotactic body radiation therapy as definitive local therapy, including dose and fractionation. CONCLUSIONS Presently, data regarding clinical benefits of local therapy on overall and other survival outcomes is still sparse for oligometastatic NSCLC. However, with rapidly evolving data being generated supporting local therapy in oligometastatic NSCLC, this guideline attempted to frame recommendations as a function of the quality of data available to make decisions in a multidisciplinary approach incorporating patient goals and tolerances.
Collapse
Affiliation(s)
- Puneeth Iyengar
- Department of Radiation Oncology, UT Southwestern, Dallas, Texas.
| | - Sean All
- Department of Radiation Oncology, UT Southwestern, Dallas, Texas
| | - Mark F Berry
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, California
| | - Thomas P Boike
- Department of Radiation Oncology, GenesisCare/MHP Radiation Oncology, Troy, Michigan
| | - Lisa Bradfield
- American Society for Radiation Oncology, Arlington, Virginia
| | - Anne-Marie C Dingemans
- Department of Pulmonology, Erasmus Medical Center Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | | | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul J Hesketh
- Department of Internal Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Melenda Jeter
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - Yolande Lievens
- Department of Radiation Oncology, Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Fiona McDonald
- Department of Radiation Oncology, Royal Marsden Hospital, London, United Kingdom
| | - Bradford A Perez
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | - Enrico Ruffini
- Department of Thoracic Surgery, University of Torino, Torino, Italy
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht and Erasmus Medical Center, University Medical Center, Rotterdam, The Netherlands
| | - Hina Saeed
- Department of Radiation Oncology, Baptist Health South Florida, Boca Raton, Florida
| | - Bryan J Schneider
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Suresh Senan
- Department of Radiation Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Joachim Widder
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Katipally RR, Martinez CA, Pugh SA, Bridgewater JA, Primrose JN, Domingo E, Maughan TS, Talamonti MS, Posner MC, Weichselbaum RR, Pitroda SP. Integrated Clinical-Molecular Classification of Colorectal Liver Metastases: A Biomarker Analysis of the Phase 3 New EPOC Randomized Clinical Trial. JAMA Oncol 2023; 9:1245-1254. [PMID: 37471075 PMCID: PMC10360005 DOI: 10.1001/jamaoncol.2023.2535] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/09/2023] [Indexed: 07/21/2023]
Abstract
Importance Personalized treatment approaches for patients with oligometastatic colorectal liver metastases are critically needed. We previously defined 3 biologically distinct molecular subtypes of colorectal liver metastases: (1) canonical, (2) immune, and (3) stromal. Objective To independently validate these molecular subtypes in the phase 3 New EPOC randomized clinical trial. Design, Setting, and Participants This retrospective secondary analysis of the phase 3 New EPOC randomized clinical trial included a bi-institutional discovery cohort and multi-institutional validation cohort. The discovery cohort comprised patients who underwent hepatic resection for limited colorectal liver metastases (98% received perioperative chemotherapy) from May 31, 1994, to August 14, 2012. The validation cohort comprised patients who underwent hepatic resection for liver metastases with perioperative chemotherapy (fluorouracil, oxaliplatin, and irinotecan based) with or without cetuximab from February 26, 2007, to November 1, 2012. Data were analyzed from January 18 to December 10, 2021. Interventions Resected metastases underwent RNA sequencing and microRNA (miRNA) profiling in the discovery cohort and messenger RNA and miRNA profiling with microarray in the validation cohort. Main Outcomes and Measures A 31-feature (24 messenger RNAs and 7 miRNAs) neural network classifier was trained to predict molecular subtypes in the discovery cohort and applied to the validation cohort. Integrated clinical-molecular risk groups were designated based on molecular subtypes and the clinical risk score. The unique biological phenotype of each molecular subtype was validated using gene set enrichment analyses and immune deconvolution. The primary clinical end points were progression-free survival (PFS) and overall survival (OS). Results A total of 240 patients were included (mean [range] age, 63.0 [56.3-68.0] years; 151 [63%] male), with 93 in the discovery cohort and 147 in the validation cohort. In the validation cohort, 73 (50%), 28 (19%), and 46 (31%) patients were classified as having canonical, immune, and stromal metastases, respectively. The biological phenotype of each subtype was concordant with the discovery cohort. The immune subtype (best prognosis) demonstrated 5-year PFS of 43% (95% CI, 25%-60%; hazard ratio [HR], 0.37; 95% CI, 0.20-0.68) and OS of 63% (95% CI, 40%-79%; HR, 0.38; 95% CI, 0.17-0.86), which was statistically significantly higher than the canonical subtype (worst prognosis) at 14% (95% CI, 7%-23%) and 43% (95% CI, 32%-55%), respectively. Adding molecular subtypes to the clinical risk score improved prediction (the Gönen and Heller K for discrimination) from 0.55 (95% CI, 0.49-0.61) to 0.62 (95% CI, 0.57-0.67) for PFS and 0.59 (95% CI, 0.52-0.66) to 0.63 (95% CI, 0.56-0.70) for OS. The low-risk integrated group demonstrated 5-year PFS of 44% (95% CI, 20%-66%; HR, 0.38; 95% CI, 0.19-0.76) and OS of 78% (95% CI, 44%-93%; HR, 0.26; 95% CI, 0.08-0.84), superior to the high-risk group at 16% (95% CI, 10%-24%) and 43% (95% CI, 32%-52%), respectively. Conclusions and Relevance In this prognostic study, biologically derived colorectal liver metastasis molecular subtypes and integrated clinical-molecular risk groups were highly prognostic. This novel molecular classification warrants further study as a possible predictive biomarker for personalized systemic treatment for colorectal liver metastases. Trial Registration isrctn.org Identifier: ISRCTN22944367.
Collapse
Affiliation(s)
- Rohan R. Katipally
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Carlos A. Martinez
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Siân A. Pugh
- Department of Oncology, Addenbrooke’s Hospital, Cambridge, England, United Kingdom
| | - John A. Bridgewater
- UCL Cancer Institute, University College London, London, England, United Kingdom
| | - John N. Primrose
- Department of Surgery, University of Southampton, Southampton, England, United Kingdom
| | - Enric Domingo
- Department of Oncology, University of Oxford, Oxford, England, United Kingdom
| | - Timothy S. Maughan
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, England, United Kingdom
| | - Mark S. Talamonti
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Mitchell C. Posner
- Department of Surgery, The University of Chicago Medicine, Chicago, Illinois
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | | |
Collapse
|
24
|
Baboudjian M, Roubaud G, Fromont G, Gauthé M, Beauval JB, Barret E, Brureau L, Créhange G, Dariane C, Fiard G, Mathieu R, Ruffion A, Rouprêt M, Renard-Penna R, Sargos P, Ploussard G. What is the ideal combination therapy in de novo, oligometastatic, castration-sensitive prostate cancer? World J Urol 2023; 41:2033-2041. [PMID: 36484817 DOI: 10.1007/s00345-022-04239-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To review current evidence regarding the management of de novo, oligometastatic, castration-sensitive prostate cancer (PCa). METHODS A literature search was conducted on PubMed/Medline and a narrative synthesis of the evidence was performed in August 2022. RESULTS Oligometastatic disease is an intermediate state between localized and aggressive metastatic PCa defined by ≤ 3-5 metastatic lesions, although this definition remains controversial. Conventional imaging has limited accuracy in detecting metastatic lesions, and the implementation of molecular imaging could pave the way for a more personalized treatment strategy. However, oncological data supporting this strategy are needed. Radiotherapy to the primary tumor should be considered standard treatment for oligometastatic PCa (omPCa). However, it remains to be seen whether local therapy still has an additional survival benefit in patients with de novo omPCa when treated with the most modern systemic therapy combinations. There is insufficient evidence to recommend cytoreductive radical prostatectomy as local therapy; or stereotactic body radiotherapy as metastasis-directed therapy in patients with omPCa. Current data support the use of intensified systemic therapy with androgen deprivation therapy (ADT) and next-generation hormone therapies (NHT) for patients with de novo omPCa. Docetaxel has not demonstrated benefit in low volume disease. There are insufficient data to support the use of triple therapy (i.e., ADT + NHT + Docetaxel) in low volume disease. CONCLUSION The present review discusses current data in de novo, omPCa regarding its definition, the increasing role of molecular imaging, the place of local and metastasis-directed therapies, and the intensification of systemic therapies.
Collapse
Affiliation(s)
- Michael Baboudjian
- Department of Urology, APHM, North Academic Hospital, Marseille, France.
- Department of Urology, APHM, La Conception Hospital, Marseille, France.
- Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain.
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, France.
| | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 33000, Bordeaux, France
| | | | - Mathieu Gauthé
- Department of Nuclear Medicine, Scintep-Institut Daniel Hollard, Grenoble, France
| | | | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | - Laurent Brureau
- Department of Urology, CHU de Pointe-à-Pitre, University of Antilles, University of Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, 97110, Pointe-à-Pitre, France
| | | | - Charles Dariane
- Department of Urology, Hôpital Européen Georges-Pompidou, APHP, Paris-Paris University-U1151 Inserm-INEM, Necker, Paris, France
| | - Gaëlle Fiard
- Department of Urology, Grenoble Alpes University Hospital, Université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France
| | | | - Alain Ruffion
- Service d'urologie Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
- Equipe 2-Centre d'Innovation en Cancérologie de Lyon (EA 3738 CICLY)-Faculté de Médecine Lyon Sud-Université Lyon 1, Lyon, France
| | - Morgan Rouprêt
- Sorbonne University, GRC 5 Predictive Onco-Uro, AP-HP, Urology, Pitie-Salpetriere Hospital, 75013, Paris, France
| | - Raphaële Renard-Penna
- Sorbonne University, AP-HP, Radiology, Pitie-Salpetriere Hospital, 75013, Paris, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, 33000, Bordeaux, France
| | - Guillaume Ploussard
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, France
- Department of Urology, Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| |
Collapse
|
25
|
Katipally RR, Pitroda SP, Weichselbaum RR, Hellman S. Oligometastases: Characterizing the Role of Epigenetic Regulation of Epithelial-Mesenchymal Transition. Clin Cancer Res 2023; 29:2761-2766. [PMID: 37115507 PMCID: PMC10687742 DOI: 10.1158/1078-0432.ccr-23-0376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
The "oligometastasis" hypothesis proposes that metastases exist as a spectrum and are not always disseminated. According to this theory, a subset of patients with metastatic disease could benefit from aggressive local therapies. However, the identification of patients most likely to exhibit an oligometastatic phenotype remains challenging. Recent literature focusing on basic and translational studies has identified novel epigenetic regulators of epithelial-mesenchymal transition (EMT) and the emergence of a spectrum of metastatic behavior. Herein, we review these scientific advances and suggest that the spectrum of metastatic virulence produced by these epigenetic mechanisms broadly contributes to the emergence of clinically evident "oligometastases." Epigenetic regulation of EMT programs can result in a spectrum of cell trajectories (e.g., quasi-mesenchymal and highly mesenchymal states) with differential propensity to develop metastases. We propose that quasi-mesenchymal cell states may be associated with a polymetastatic phenotype, whereas highly mesenchymal cell states may be associated with a more oligometastatic phenotype. The mechanisms governing epigenetic regulation of EMT and its array of intermediate states are multifaceted and may contribute to the development of the metastatic spectrum observed clinically. Within this context, translational studies that support the role of EMT and its epigenetic regulation are discussed. Continued translation of these mechanistic discoveries into novel biomarkers may help optimally select patients most likely to exhibit an oligometastatic phenotype and benefit from aggressive local therapies, such as surgery, radiotherapy, and other ablative procedures.
Collapse
Affiliation(s)
- Rohan R. Katipally
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL, USA
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL, USA
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL, USA
| | - Samuel Hellman
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
26
|
He X, Yeung SJ, Esteva FJ. A new paradigm for classifying and treating HER2-positive breast cancer. Cancer Rep (Hoboken) 2023; 6:e1841. [PMID: 37254964 PMCID: PMC10432420 DOI: 10.1002/cnr2.1841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Because of the phenomenal success of treatment with monoclonal antibodies and antibody-drug conjugates targeting human epidermal growth factor receptor 2 (HER2), most patients with early-stage HER2-positive breast cancer (HER2+ BC) and some with limited metastatic diseases have been cured, and those who have not been cured have achieved significant improvements in overall survival, which has weakened the role of the TNM staging system in the prognosis of HER2+ BC today. Given that the disease is now highly curable, treatment conception, classification, and modalities should differ from those of cancer types with a poor prognosis. It is warranted to build a new paradigm for classifying and treating HER2+ BC. RECENT FINDINGS In our personal view, we suggest that the classification system should be based not only on traditional anatomy and cancer biology but also on available treatment regimens, their exceptional outcomes, and their toxicities. In this regard, we developed a new concise classification of HER2+ BC based on the TNM staging system, a review of the literature, research results, and our clinical experience, dividing the patients into four distinct groups: curable (lymph-node negative and small (≤3 cm) early breast cancer), do our best to cure (locally advanced or tumors >3 cm early breast cancer), hope for cure (local-regional recurrent, limited metastases, and exceptional responders), and incurable (metastatic breast cancer with poor performance status or non-exceptional responders). CONCLUSION It will assist clinicians in developing an optimal treatment regimen at the outset, curing more HER2+ BC patients and improving their quality of life.
Collapse
Affiliation(s)
- Xuexin He
- Department of Medical OncologyHuashan Hospital of Fudan UniversityShanghaiChina
| | | | - Francisco J. Esteva
- Division of Hematology/OncologyNorthwell Health Cancer Institute at Lenox Hill HospitalNew YorkNew YorkUSA
| |
Collapse
|
27
|
Zhou ML, Xu RN, Tan C, Zhang Z, Wan JF. Advanced gastric cancer achieving major pathologic regression after chemoimmunotherapy combined with hypofractionated radiotherapy: A case report. World J Gastrointest Oncol 2023; 15:1096-1104. [PMID: 37389115 PMCID: PMC10302995 DOI: 10.4251/wjgo.v15.i6.1096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Currently, chemotherapy combined with immunotherapy is the established first-line standard treatment for advanced gastric cancer (GC). In addition, the combination of radiotherapy and immunotherapy is considered a promising treatment strategy.
CASE SUMMARY In this report, we present a case of achieving nearly complete remission of highly advanced GC with comprehensive therapies. A 67-year-old male patient was referred to the hospital because he presented with dyspepsia and melena for several days. Based on fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT), endoscopic examination and abdominal CT, he was diagnosed with GC with a massive lesion and two distant metastatic lesions. The patient received mFOLFOX6 regimen chemotherapy, nivolumab and a short course of hypofractionated radiotherapy (4 Gy × 6 fractions) targeting the primary lesion. After the completion of these therapies, the tumor and the metastatic lesions showed a partial response. After having this case discussed by a multidisciplinary team, the patient underwent surgery, including total gastrectomy and D2 lymph node dissection. Postoperative pathology showed that major pathological regression of the primary lesion was achieved. Chemoimmunotherapy started four weeks after surgery, and examination was performed every three months. Since surgery, the patient has been stable and healthy with no evidence of recurrence.
CONCLUSION The combination of radiotherapy and immunotherapy for GC is worthy of further exploration.
Collapse
Affiliation(s)
- Meng-Long Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Ruo-Ne Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Cong Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Jue-Feng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| |
Collapse
|
28
|
Li D, Xu W, Chang Y, Xiao Y, He Y, Ren S. Advances in landscape and related therapeutic targets of the prostate tumor microenvironment. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37294106 DOI: 10.3724/abbs.2023092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
The distinct tumor microenvironment (TME) of prostate cancer (PCa), which promotes tumor proliferation and progression, consists of various stromal cells, immune cells, and a dense extracellular matrix (ECM). The understanding of the prostate TME extends to tertiary lymphoid structures (TLSs) and metastasis niches to provide a more concise comprehension of tumor metastasis. These constituents collectively structure the hallmarks of the pro-tumor TME, including immunosuppressive, acidic, and hypoxic niches, neuronal innervation, and metabolic rewiring. In combination with the knowledge of the tumor microenvironment and the advancement of emerging therapeutic technologies, several therapeutic strategies have been developed, and some of them have been tested in clinical trials. This review elaborates on PCa TME components, summarizes various TME-targeted therapies, and provides insights into PCa carcinogenesis, progression, and therapeutic strategies.
Collapse
Affiliation(s)
- Duocai Li
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Weidong Xu
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yifan Chang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yutian Xiao
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
29
|
Tang C, Sherry AD, Haymaker C, Bathala T, Liu S, Fellman B, Cohen L, Aparicio A, Zurita AJ, Reuben A, Marmonti E, Chun SG, Reddy JP, Ghia A, McGuire S, Efstathiou E, Wang J, Wang J, Pilie P, Kovitz C, Du W, Simiele SJ, Kumar R, Borghero Y, Shi Z, Chapin B, Gomez D, Wistuba I, Corn PG. Addition of Metastasis-Directed Therapy to Intermittent Hormone Therapy for Oligometastatic Prostate Cancer: The EXTEND Phase 2 Randomized Clinical Trial. JAMA Oncol 2023; 9:825-834. [PMID: 37022702 PMCID: PMC10080407 DOI: 10.1001/jamaoncol.2023.0161] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/20/2022] [Indexed: 04/07/2023]
Abstract
Importance Despite evidence demonstrating an overall survival benefit with up-front hormone therapy in addition to established synergy between hormone therapy and radiation, the addition of metastasis-directed therapy (MDT) to hormone therapy for oligometastatic prostate cancer, to date, has not been evaluated in a randomized clinical trial. Objective To determine in men with oligometastatic prostate cancer whether the addition of MDT to intermittent hormone therapy improves oncologic outcomes and preserves time with eugonadal testosterone compared with intermittent hormone therapy alone. Design, Setting, Participants The External Beam Radiation to Eliminate Nominal Metastatic Disease (EXTEND) trial is a phase 2, basket randomized clinical trial for multiple solid tumors testing the addition of MDT to standard-of-care systemic therapy. Men aged 18 years or older with oligometastatic prostate cancer who had 5 or fewer metastases and were treated with hormone therapy for 2 or more months were enrolled to the prostate intermittent hormone therapy basket at multicenter tertiary cancer centers from September 2018 to November 2020. The cutoff date for the primary analysis was January 7, 2022. Interventions Patients were randomized 1:1 to MDT, consisting of definitive radiation therapy to all sites of disease and intermittent hormone therapy (combined therapy arm; n = 43) or to hormone therapy only (n = 44). A planned break in hormone therapy occurred 6 months after enrollment, after which hormone therapy was withheld until progression. Main Outcomes and Measures The primary end point was disease progression, defined as death or radiographic, clinical, or biochemical progression. A key predefined secondary end point was eugonadal progression-free survival (PFS), defined as the time from achieving a eugonadal testosterone level (≥150 ng/dL; to convert to nanomoles per liter, multiply by 0.0347) until progression. Exploratory measures included quality of life and systemic immune evaluation using flow cytometry and T-cell receptor sequencing. Results The study included 87 men (median age, 67 years [IQR, 63-72 years]). Median follow-up was 22.0 months (range, 11.6-39.2 months). Progression-free survival was improved in the combined therapy arm (median not reached) compared with the hormone therapy only arm (median, 15.8 months; 95% CI, 13.6-21.2 months) (hazard ratio, 0.25; 95% CI, 0.12-0.55; P < .001). Eugonadal PFS was also improved with MDT (median not reached) compared with the hormone therapy only (6.1 months; 95% CI, 3.7 months to not estimable) (hazard ratio, 0.32; 95% CI, 0.11-0.91; P = .03). Flow cytometry and T-cell receptor sequencing demonstrated increased markers of T-cell activation, proliferation, and clonal expansion limited to the combined therapy arm. Conclusions and Relevance In this randomized clinical trial, PFS and eugonadal PFS were significantly improved with combination treatment compared with hormone treatment only in men with oligometastatic prostate cancer. Combination of MDT with intermittent hormone therapy may allow for excellent disease control while facilitating prolonged eugonadal testosterone intervals. Trial Registration ClinicalTrials.gov Identifier: NCT03599765.
Collapse
Affiliation(s)
- Chad Tang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston
| | - Alexander D. Sherry
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Tharakeswara Bathala
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Lorenzo Cohen
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Amado J. Zurita
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Enrica Marmonti
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Stephen G. Chun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jay P. Reddy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Amol Ghia
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Sean McGuire
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jianbo Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Patrick Pilie
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Craig Kovitz
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Weiliang Du
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston
| | - Samantha J. Simiele
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston
| | - Rachit Kumar
- Department of Radiation Oncology, Banner MD Anderson Cancer Center, Gilbert, Arizona
| | - Yerko Borghero
- Department of Radiation Oncology, Banner MD Anderson Cancer Center, Gilbert, Arizona
| | - Zheng Shi
- Department of Radiation Oncology, The University of Texas Health Science Center at San Antonio
| | - Brian Chapin
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Paul G. Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
30
|
Meziani L, Gerbé de Thoré M, Clémenson C, Liu W, Laurent PA, Mondini M, Vozenin MC, Deutsch E. Optimal dosing regimen of CD73 blockade improves tumor response to radiotherapy through iCOS downregulation. J Immunother Cancer 2023; 11:jitc-2023-006846. [PMID: 37270182 DOI: 10.1136/jitc-2023-006846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Irradiation (IR) and immune checkpoint inhibitor (ICI) combination is a promising treatment modality. However, local and distance treatment failure and resistance can occur. To counteract this resistance, several studies propose CD73, an ectoenzyme, as a potential target to improve the antitumor efficiency of IR and ICI. Although CD73 targeting in combination with IR and ICI has shown attractive antitumor effects in preclinical models, the rationale for CD73 targeting based on CD73 tumor expression level deserves further investigations. METHODS Here we evaluated for the first time the efficacy of two administration regimens of CD73 neutralizing antibody (one dose vs four doses) in combination with IR according to the expression level of CD73 in two subcutaneous tumor models expressing different levels of CD73. RESULTS We showed that CD73 is weakly expressed by MC38 tumors even after IR, when compared with the TS/A model that highly expressed CD73. Treatment with four doses of anti-CD73 improved the TS/A tumor response to IR, while it was ineffective against the CD73 low-expressing MC38 tumors. Surprisingly, a single dose of anti-CD73 exerted a significant antitumor activity against MC38 tumors. On CD73 overexpression in MC38 cells, four doses of anti-CD73 were required to improve the efficacy of IR. Mechanistically, a correlation between a downregulation of iCOS expression in CD4+ T cells and an improved response to IR after anti-CD73 treatment was observed and iCOS targeting could restore an impaired benefit from anti-CD73 treatment. CONCLUSIONS These data emphasize the importance of the dosing regimen for anti-CD73 treatment to improve tumor response to IR and identify iCOS as part of the underlying molecular mechanisms. Our data suggest that the selection of appropriate dosing regimen is required to optimize the therapeutic efficacy of immunotherapy-radiotherapy combinations.
Collapse
Affiliation(s)
- Lydia Meziani
- Laboratory of Radiation Oncology, Department of Radiation Oncology, CHUV, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Marine Gerbé de Thoré
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Céline Clémenson
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Winchygn Liu
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Pierre-Antoine Laurent
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Michele Mondini
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, CHUV, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric Deutsch
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
31
|
Song M, Zeng X, Wu Q, Huang J, Dong J, Shao L, Sun Z, Lin Y, Chen S. Metastatic Colorectal Cancer Patient with Microsatellite Stability and Germline BRAC2 Mutation Shows a Complete Response to Olaparib in Combination with a PD-1 Inhibitor and Bevacizumab: A Case Report and Review of the Literature. Life (Basel) 2023; 13:life13051183. [PMID: 37240828 DOI: 10.3390/life13051183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Metastatic colorectal cancer (mCRC) has a poor prognosis. Combining chemotherapy with targeted therapy constitutes a basic form of mCRC treatment. Immune checkpoint inhibitors have been recommended for microsatellite instability mCRC, while most patients harboring microsatellite stability (MSS) or proficient mismatch repair (pMMR) are less responsive to immunotherapy. Combinational targeted therapy, including poly-ADP ribose polymerase (PARP) inhibitors, has been considered a promising way to reverse immunotherapy resistance; however, there is no clear and consistent conclusions can be drawn from the current research. Here, we report the case of a 59-year-old woman diagnosed with stage IVB MSS mCRC who received three courses of capecitabine/oxaliplatin chemotherapy combined with bevacizumab as a first-line treatment, resulting in an overall evaluation of stable disease (-25.7%). However, the occurrence of adverse events of intolerable grade 3 diarrhea and vomiting forced the cessation of this therapy. A germline BRCA2 mutation was found by next-generation sequencing, and the patient further received a combination of olaparib, tislelizumab, and bevacizumab. This treatment regime resulted in a complete metabolic response and a partial response (-50.9%) after 3 months of treatment. Mild asymptomatic interstitial pneumonia and manageable hematologic toxicity were two adverse events associated with this combination therapy. This study provides new insights into the combination of PARP inhibitors and immunotherapy for MSS mCRC patients carrying germline BRCA2 mutations.
Collapse
Affiliation(s)
- Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xianrong Zeng
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Qian Wu
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Jie Huang
- Department of Oncology, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
| | - Jiayi Dong
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Lijuan Shao
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Zihao Sun
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Yiguang Lin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangzhou Anjie Biomedical Technology Co., Ltd., Guangzhou 510530, China
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| |
Collapse
|
32
|
Liu SYM, Zheng MM, Pan Y, Liu SY, Li Y, Wu YL. Emerging evidence and treatment paradigm of non-small cell lung cancer. J Hematol Oncol 2023; 16:40. [PMID: 37069698 PMCID: PMC10108547 DOI: 10.1186/s13045-023-01436-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
Research on biomarker-driven therapy and immune check-point blockade in non-small cell lung cancer (NSCLC) is rapidly evolving. The width and depth of clinical trials have also dramatically improved in an unprecedented speed. The personalized treatment paradigm evolved every year. In this review, we summarize the promising agents that have shifted the treatment paradigm for NSCLC patients across all stages, including targeted therapy and immunotherapy using checkpoint inhibitors. Based on recent evidence, we propose treatment algorithms for NSCLC and propose several unsolved clinical issues, which are being explored in ongoing clinical trials. The results of these trials are likely to impact future clinical practice.
Collapse
Affiliation(s)
- Si-Yang Maggie Liu
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Mei-Mei Zheng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yi Pan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yangqiu Li
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Gerstberger S, Jiang Q, Ganesh K. Metastasis. Cell 2023; 186:1564-1579. [PMID: 37059065 PMCID: PMC10511214 DOI: 10.1016/j.cell.2023.03.003] [Citation(s) in RCA: 259] [Impact Index Per Article: 129.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/16/2023] [Accepted: 03/02/2023] [Indexed: 04/16/2023]
Abstract
Most cancer-associated deaths occur due to metastasis, yet our understanding of metastasis as an evolving, heterogeneous, systemic disease and of how to effectively treat it is still emerging. Metastasis requires the acquisition of a succession of traits to disseminate, variably enter and exit dormancy, and colonize distant organs. The success of these events is driven by clonal selection, the potential of metastatic cells to dynamically transition into distinct states, and their ability to co-opt the immune environment. Here, we review the main principles of metastasis and highlight emerging opportunities to develop more effective therapies for metastatic cancer.
Collapse
Affiliation(s)
- Stefanie Gerstberger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qingwen Jiang
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karuna Ganesh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
34
|
Lacaze JL, Chira C, Glemarec G, Monselet N, Cassou-Mounat T, De Maio E, Jouve E, Massabeau C, Brac de la Perrière C, Selmes G, Ung M, Nicolai V, Cabarrou B, Dalenc F. Clinical and pathological characterization of 158 consecutive and unselected oligometastatic breast cancers in a single institution. Breast Cancer Res Treat 2023; 198:463-474. [PMID: 36790573 DOI: 10.1007/s10549-023-06880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
PURPOSE Data about incidence, biological, and clinical characteristics of oligometastatic breast cancer (OMBC) are scarce. However, these data are essential in determining optimal treatment strategy. Gaining knowledge of these elements means observing and describing large, recent, and consecutive series of OMBC in their natural history. METHODS We collected data retrospectively at our institution from 998 consecutive patients diagnosed and treated with synchronous or metachronous metastatic breast cancer (MBC) between January 2014 and December 2018. The only criterion used to define OMBC was the presence of one to five metastases at diagnosis. RESULTS Of 998 MBC, 15.8% were classified OMBC. Among these, 88% had one to three metastases, and 86.7% had only one organ involved. Bone metastases were present in 52.5% of cases, 20.9% had progression to lymph nodes, 14.6% to the liver, 13.3% to the brain, 8.2% to the lungs, and 3.8% had other metastases. 55.7% had HR+/HER2- OMBC, 25.3% had HER2+OMBC, and 19% had HR-/HER2- OMBC. The HR+/HER2- subtype statistically correlated with bone metastases (p = 0.001), the HER2+subtype with brain lesions (p = 0.001), and the HR-/HER2- subtype with lymph node metastases (p = 0.008). Visceral metastases were not statistically associated with any OMBC subtypes (p = 0.186). OMBC-SBR grade III was proportionally higher than in the ESME series of 22,109 MBC (49.4% vs. 35.1%, p < 0.001). CONCLUSION OMBC is a heterogeneous entity whose incidence is higher than has commonly been published. Not an indolent disease, each subgroup, with its biological and anatomical characteristics, merits specific management.
Collapse
Affiliation(s)
- Jean-Louis Lacaze
- Départment d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France.
| | - Ciprian Chira
- Département de Radiothérapie, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Gauthier Glemarec
- Département de Radiothérapie, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Nils Monselet
- Biostatistics & Health Data Science Unit, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Thibaut Cassou-Mounat
- Département de Médecine Nucléaire, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Eleonora De Maio
- Départment d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Eva Jouve
- Département de Chirurgie, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Carole Massabeau
- Département de Radiothérapie, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Clémence Brac de la Perrière
- Départment d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Gabrielle Selmes
- Département de Chirurgie, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Mony Ung
- Départment d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Vincent Nicolai
- Départment d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Bastien Cabarrou
- Biostatistics & Health Data Science Unit, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), 1 Av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| | - Florence Dalenc
- Département d'Oncologie Médicale, Institut Claudius Regaud (ICR), Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, 1 av. Irène Joliot Curie, 31059, Toulouse Cedex 9, France
| |
Collapse
|
35
|
Hardy NP, Epperlein JP, Dalli J, Robertson W, Liddy R, Aird JJ, Mulligan N, Neary PM, McEntee GP, Conneely JB, Cahill RA. Real-time administration of indocyanine green in combination with computer vision and artificial intelligence for the identification and delineation of colorectal liver metastases. Surg Open Sci 2023; 12:48-54. [PMID: 36936453 PMCID: PMC10017420 DOI: 10.1016/j.sopen.2023.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Fluorescence guided surgery for the identification of colorectal liver metastases (CRLM) can be better with low specificity and antecedent dosing impracticalities limiting indocyanine green (ICG) usefulness currently. We investigated the application of artificial intelligence methods (AIM) to demonstrate and characterise CLRMs based on dynamic signalling immediately following intraoperative ICG administration. Methods Twenty-five patients with liver surface lesions (24 CRLM and 1 benign cyst) undergoing open/laparoscopic/robotic procedures were studied. ICG (0.05 mg/kg) was administered with near-infrared recording of fluorescence perfusion. User-selected region-of-interest (ROI) perfusion profiles were generated, milestones relating to ICG inflow/outflow extracted and used to train a machine learning (ML) classifier. 2D heatmaps were constructed in a subset using AIM to depict whole screen imaging based on dynamic tissue-ICG interaction. Fluorescence appearances were also assessed microscopically (using H&E and fresh-frozen preparations) to provide tissue-level explainability of such methods. Results The ML algorithm correctly classified 97.2 % of CRLM ROIs (n = 132) and all benign lesion ROIs (n = 6) within 90-s of ICG administration following initial mathematical curve analysis identifying ICG inflow/outflow differentials between healthy liver and CRLMs. Time-fluorescence plots extracted for each pixel in 10 lesions enabled creation of 2D characterising heatmaps using flow parameters and through unsupervised ML. Microscopy confirmed statistically less CLRM fluorescence vs adjacent liver (mean ± std deviation signal/area 2.46 ± 9.56 vs 507.43 ± 160.82 respectively p < 0.001) with H&E diminishing ICG signal (n = 4). Conclusion ML accurately identifies CRLMs from surrounding liver tissue enabling representative 2D mapping of such lesions from their fluorescence perfusion patterns using AIM. This may assist in reducing positive margin rates at metastatectomy and in identifying unexpected/occult malignancies.
Collapse
Affiliation(s)
- Niall P. Hardy
- UCD Centre for Precision Surgery, School of Medicine, UCD, Dublin, Ireland
| | | | - Jeffrey Dalli
- UCD Centre for Precision Surgery, School of Medicine, UCD, Dublin, Ireland
| | - William Robertson
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Richard Liddy
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - John J. Aird
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Niall Mulligan
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Peter M. Neary
- Department of General and Colorectal Surgery, University Hospital Waterford, University College Cork, Ireland
| | - Gerard P. McEntee
- Department of Hepatobiliary, Foregut and Bariatric Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - John B. Conneely
- Department of Hepatobiliary, Foregut and Bariatric Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ronan A. Cahill
- UCD Centre for Precision Surgery, School of Medicine, UCD, Dublin, Ireland
- Department of General and Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
- Corresponding author at: 47 Eccles Street, Dublin 7, Ireland. @Matersurgery
| |
Collapse
|
36
|
Bordeau K, Michalet M, Keskes A, Valdenaire S, Debuire P, Cantaloube M, Cabaillé M, Jacot W, Draghici R, Demontoy S, Quantin X, Ychou M, Assenat E, Mazard T, Gauthier L, Dupuy M, Guiu B, Bourgier C, Aillères N, Fenoglietto P, Azria D, Riou O. Stereotactic MR-Guided Radiotherapy for Liver Metastases: First Results of the Montpellier Prospective Registry Study. J Clin Med 2023; 12:jcm12031183. [PMID: 36769831 PMCID: PMC9917771 DOI: 10.3390/jcm12031183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Liver stereotactic body radiotherapy (SBRT) is a local treatment that provides good local control and low toxicity. We present the first clinical results from our prospective registry of stereotactic MR-guided radiotherapy (MRgRT) for liver metastases. All patients treated for liver metastases were included in this prospective registry study. Stereotactic MRgRT indication was confirmed by multidisciplinary specialized tumor boards. The primary endpoints were acute and late toxicities. The secondary endpoints were survival outcomes (local control, overall survival (OS), disease-free survival, intrahepatic relapse-free survival). Twenty-six consecutive patients were treated for thirty-one liver metastases between October 2019 and April 2022. The median prescribed dose was 50 Gy (40-60) in 5 fractions. No severe acute MRgRT-related toxicity was noted. Acute and late gastrointestinal and liver toxicities were low and mostly unrelated to MRgRT. Only 5 lesions (16.1%) required daily adaptation because of the proximity of organs at risk (OAR). With a median follow-up time of 17.3 months since MRgRT completion, the median OS, 1-year OS and 2-year OS rates were 21.7 months, 83.1% (95% CI: 55.3-94.4%) and 41.6% (95% CI: 13.5-68.1%), respectively, from MRgRT completion. The local control at 6 months, 1 year and 2 years was 90.9% (95% CI: 68.3-97.7%). To our knowledge, we report the largest series of stereotactic MRgRT for liver metastases. The treatment was well-tolerated and achieved a high LC rate. Distant relapse remains a challenge in this population.
Collapse
Affiliation(s)
- Karl Bordeau
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Morgan Michalet
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Aïcha Keskes
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Simon Valdenaire
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Pierre Debuire
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Marie Cantaloube
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Morgane Cabaillé
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - William Jacot
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University Montpellier, 34298 Montpellier, France
| | - Roxana Draghici
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Sylvain Demontoy
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Xavier Quantin
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University Montpellier, 34298 Montpellier, France
| | - Marc Ychou
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University Montpellier, 34298 Montpellier, France
| | - Eric Assenat
- Medical Oncology Department, CHU St. Eloi, 34000 Montpellier, France
| | - Thibault Mazard
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University Montpellier, 34298 Montpellier, France
| | - Ludovic Gauthier
- Biometrics Unit, Montpellier Cancer Institute (ICM), University Montpellier, 34298 Montpellier, France
| | - Marie Dupuy
- Medical Oncology Department, CHU St. Eloi, 34000 Montpellier, France
| | - Boris Guiu
- Radiology Department, CHU St. Eloi, 34000 Montpellier, France
| | - Céline Bourgier
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Norbert Aillères
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Pascal Fenoglietto
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - David Azria
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
| | - Olivier Riou
- Montpellier Cancer Institute (ICM), University Federation of Radiation Oncology of Mediterranean Occitanie, University Montpellier, INSERM U1194 IRCM, 34298 Montpellier, France
- Correspondence:
| |
Collapse
|
37
|
Pérez-Montero H, Lozano A, de Blas R, Sánchez JJ, Martínez E, Laplana M, Gil-Gil M, Garcia-Tejedor A, Pernas S, Falo C, Godino Ó, Pla MJ, Guedea F, Navarro-Martin A. Ten-year experience of bone SBRT in breast cancer: analysis of predictive factors of effectiveness. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1756-1766. [PMID: 36645616 DOI: 10.1007/s12094-023-03073-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/03/2023] [Indexed: 01/17/2023]
Abstract
PURPOSE Data on the benefit of stereotactic body radiation therapy (SBRT) in patients with breast cancer (BC) and bone metastases remain limited. The purpose of this study is to report our 10-year experience of bone SBRT, analyzing toxicity and prognostic factors for local control (LC); progression-free survival, and overall survival (OS). METHODS/PATIENTS We analyzed all spine and non-spine bone SBRT performed in patients with BC during the 2012-2022 period at our institution. Treatments carried out with ablative intent in stereotactic conditions with dose/fraction ≥ 5 Gy in 5 or fewer sessions were considered. Demographic, treatment, and toxicity data were recorded according to CTCAEv4. Risk factors were assessed through univariate and multivariate analysis by Cox regression. RESULTS 60 bone SBRT treatments were performed during the study period. 75% were spine SBRT and 25% were non-spine SBRT (median BED4Gy was 80 Gy4). The median age was 52.5 years (34-79). The median tumor volume was 2.9 cm3 (0.5-39.4). The median follow-up was 32.4 months (1.2-101.7). 1 and 2 years LC were 92.9 and 86.6%, respectively. 1 and 2 years OS were 100 and 90.6%, respectively. Multivariate analysis (MVA) associated volume of the treated lesion ≥ 13 cm3 with worse LC (p = 0.046; HR 12.1, 95%CI = 1.1-140.3). In addition, deferring SBRT > 3 months after lesion diagnosis to prioritize systemic treatment showed a significant benefit, improving the 2 years LC up to 96.8% vs. 67.5% for SBRT performed before this period (p = 0.031; HR 0.1, 95%CI = 0.01-0.8). Hormonal receptors, the total number of metastases, and CA15-3 value were significantly associated with OS in MVA. During follow-up, three non-spine fractures (5%) were observed. CONCLUSIONS According to our data, bone SBRT is a safe and effective technique for BC. Upfront systemic treatment before SBRT offers a benefit in LC. Therefore, SBRT should be considered after prior systemic treatment in this population.
Collapse
Affiliation(s)
- Héctor Pérez-Montero
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - Alicia Lozano
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - Rodolfo de Blas
- Medical Physics and Radiation Protection Department, Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - Juan José Sánchez
- Radiodiagnostic Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Carrer de la Feixa Llarga, S/N, 08907, Barcelona, Spain
| | - Evelyn Martínez
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - María Laplana
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - Miguel Gil-Gil
- Medical Oncology Department, Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Amparo Garcia-Tejedor
- Gynecology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Carrer de la Feixa Llarga, S/N, 08907, Barcelona, Spain
| | - Sonia Pernas
- Medical Oncology Department, Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Catalina Falo
- Medical Oncology Department, Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Óscar Godino
- Neurosurgery Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Carrer de la Feixa Llarga, S/N, 08907, Barcelona, Spain
| | - Maria J Pla
- Gynecology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Carrer de la Feixa Llarga, S/N, 08907, Barcelona, Spain
| | - Ferrán Guedea
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain
| | - Arturo Navarro-Martin
- Radiation Oncology Department, Catalan Institute of Oncology, Avinguda de la Gran Via de l'Hospitalet, L'Hospitalet de Llobregat, 199-203, 08908, Barcelona, Spain.
| |
Collapse
|
38
|
Kim KH, Ahn YC. Oligometastasis: More Lessons to Be Learned. Cancer Res Treat 2023; 55:1-4. [PMID: 36596725 PMCID: PMC9873332 DOI: 10.4143/crt.2023.265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 01/02/2023] [Indexed: 01/04/2023] Open
Affiliation(s)
- Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul,
Korea
| | - Yong Chan Ahn
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | | |
Collapse
|
39
|
Miao Q, Wei Z, Liu C, Ye Y, Cheng G, Song Z, Chen K, Zhang Y, Chen J, Yue C, Ruan H, Zhang X. Overall survival and cancer-specific survival were improved in local treatment of metastatic prostate cancer. Front Oncol 2023; 13:1130680. [PMID: 37207146 PMCID: PMC10189015 DOI: 10.3389/fonc.2023.1130680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/19/2023] [Indexed: 05/21/2023] Open
Abstract
Background For metastatic prostate cancer (mPCa), radical prostatectomy (RP) and radiation therapy (RT) may improve overall survival (OS) and cancer-specific survival (CSS). Compared with RT, RP shows significant advantages in improving patient outcomes. External beam radiation therapy (EBRT) even slightly elevates CSM with no statistical difference in OS compared with no local treatment (NLT). Objective To evaluate OS and CSS after local treatment (LT) (including RP and RT) versus NLT in mPCa. Design, setting, and participants Within the Surveillance, Epidemiology and End Results (SEER) database (2000-2018), 20098 patients with metastatic prostate cancer were selected in this study, of which 19433 patients had no local treatment, 377 patients with radical prostate treatment, and 288 patients with RT. Outcome measurements and statistical analysis Multivariable competing risks regression analysis after propensity score matching (PSM) was used to calculate CSM. Multivariable Cox regression analysis was used to identify the risk factors. Kaplan-Meier methods were used to calculate OS. Results and limitations A total of 20098 patients were included: NLT (n = 19433), RP (n=377) and RT (n=288). In a competing risk regression analysis after PSM (ratio 1:1), RP resulted in a significantly lower CSM (hazard ratio [HR] 0.36, 95% confidence interval [CI] 0.29-0.45) than NLT, while RT showed a slightly lower CSM (HR 0.77, 95% CI 0.63-0.95). In a competing risk regression analysis after PSM (ratio 1:1), RP led to a lower CSM (HR 0.56, 95% CI 0.41-0.76) versus RT. As for all-cause mortality (ACM), RP (HR 0.37, 95% CI 0.31-0.45) and RT (HR 0.66, 95% CI 0.56-0.79). also showed a downward trend. In terms of OS, RP and RT significantly improved the survival probability compared with NLT, with the effect of RP being more pronounced. Obviously, older age, Gleason scores ≥8, AJCC T3-T4 stage, AJCC N1, AJCC M1b-M1c were all associated with higher CSM (P <0.05). The same results held true for ACM. The limitation of this article is that it is not possible to assess the effect of differences in systemic therapy on CSM in mPCa patients and clinical trials are needed to verify the results. Conclusions For patients with mPCa, both RP and RT are beneficial to patients, and the efficacy of RP is better than RT from the perspective of CSM and ACM. Older age, higher gleason scores and the more advanced AJCC TNM stage all put patients at higher risk of dying. Patient summary A large population-based cancer database showed that in addition to first-line therapy (hormonal treatment), RP and radiotherapy can also benefit patients with mPCa.
Collapse
Affiliation(s)
- Qi Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengshuai Song
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunxuan Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changjie Yue
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaoping Zhang, ; Hailong Ruan,
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaoping Zhang, ; Hailong Ruan,
| |
Collapse
|
40
|
Katipally RR, Olson AC, Chmura SJ. Flipping the Null: Lessons From Breast Cancer and Recalibrating Expectations in Oligometastases. Int J Radiat Oncol Biol Phys 2022; 114:840-842. [DOI: 10.1016/j.ijrobp.2022.09.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
|
41
|
Local Therapy for Oligometastatic Disease—Cart Before the Horse? Int J Radiat Oncol Biol Phys 2022; 114:836-839. [DOI: 10.1016/j.ijrobp.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022]
|
42
|
von Deimling M, Rajwa P, Tilki D, Heidenreich A, Pallauf M, Bianchi A, Yanagisawa T, Kawada T, Karakiewicz PI, Gontero P, Pradere B, Ploussard G, Rink M, Shariat SF. The current role of precision surgery in oligometastatic prostate cancer. ESMO Open 2022; 7:100597. [PMID: 36208497 PMCID: PMC9551071 DOI: 10.1016/j.esmoop.2022.100597] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 12/30/2022] Open
Abstract
Oligometastatic prostate cancer (omPCa) is a novel intermediate disease state characterized by a limited volume of metastatic cells and specific locations. Accurate staging is paramount to unmask oligometastatic disease, as provided by prostate-specific membrane antigen-positron emission tomography. Driven by the results of prospective trials employing conventional and/or modern staging modalities, the treatment landscape of omPCa has rapidly evolved over the last years. Several treatment-related questions comprising the concept of precision strikes are under development. For example, beyond systemic therapy, cohort studies have found that cytoreductive radical prostatectomy (CRP) can confer a survival benefit in select patients with omPCa. More importantly, CRP has been consistently shown to improve long-term local symptoms when the tumor progresses across disease states due to resistance to systemic therapies. Metastasis-directed treatments have also emerged as a promising treatment option due to the visibility of oligometastatic disease and new technologies as well as treatment strategies to target the novel PCa colonies. Whether metastases are present at primary cancer diagnosis or detected upon biochemical recurrence after treatment with curative intent, targeted yet decisive elimination of disseminated tumor cell hotspots is thought to improve survival outcomes. One such strategy is salvage lymph node dissection in oligorecurrent PCa which can alter the natural history of progressive PCa. In this review, we will highlight how refinements in modern staging modalities change the classification and treatment of (oligo-)metastatic PCa. Further, we will also discuss the current role and future directions of precision surgery in omPCa.
Collapse
Affiliation(s)
- M von Deimling
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - P Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - D Tilki
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - A Heidenreich
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University Hospital Cologne, Cologne, Germany
| | - M Pallauf
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - A Bianchi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - T Yanagisawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - T Kawada
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - P I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Canada
| | - P Gontero
- Division of Urology, Department of Surgical Sciences, San Giovanni Battista Hospital, University of Studies of Torino, Turin, Italy
| | - B Pradere
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, La Croix Du Sud Hospital, Quint-Fonsegrives, France
| | - G Ploussard
- Department of Urology, La Croix Du Sud Hospital, Quint-Fonsegrives, France
| | - M Rink
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, USA; Department of Urology, University of Texas Southwestern, Dallas, USA; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| |
Collapse
|
43
|
Miglietta F, Visani L, Marini S, Griguolo G, Vernaci GM, Bottosso M, Dieci MV, Meattini I, Guarneri V. Oligometastatic breast cancer: Dissecting the clinical and biological uniqueness of this emerging entity. Can we pursue curability? Cancer Treat Rev 2022; 110:102462. [PMID: 36087503 DOI: 10.1016/j.ctrv.2022.102462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022]
Abstract
Metastatic breast cancer represents an incurable condition, however, the increasing interest towards the oligometastatic entity is now challenging this assumption. Up to 20% of patients with metastatic breast cancer present with oligometastatic disease, which refers to metastatic breast cancer presenting or recurring with limited metastatic burden. In the last years, progressive advancements in imaging techniques, the growing availability of minimally invasive locoregional treatments, alongside the increasing expectations from a patient perspective, have contributed to rising the awareness towards this emerging entity. In the present work we comprehensively reviewed available evidence regarding oligometastatic breast cancer, focusing on clinical and biological notions virtually supporting the adoption of a curative approach when treating this condition. We also discussed main areas of uncertainties, providing a research agenda that may guide and fine-tune the future investigation in this field.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Luca Visani
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Sabrina Marini
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Grazia Maria Vernaci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Michele Bottosso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Icro Meattini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| |
Collapse
|